Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Discov Med ; 36(185): 1280-1288, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38926114

RESUMEN

BACKGROUND: Kinesin family member 26B (KIF26B) has been closely linked to the occurrence and progression of various tumors. However, there is limited research on its role in oral squamous cell carcinoma (OSCC). This article aims to investigate the expression levels and mechanisms of KIF26B in OSCC. METHODS: Real time quantity polymerase chain reaction (RT-qPCR) and Western blot analyses were conducted to assess the expression levels of KIF26B in 35 OSCC specimens and their corresponding non-cancerous tissues. Overexpression and silencing of KIF26B were achieved in HSC6 and SCC25 cells, respectively, resulting in the establishment of KIF26B-overexpressing and si-KIF26B cell lines, designated as the KIF26B group and si-KIF26B group. Proliferation assays using 5-Ethynyl-2'-deoxyuridine (EdU) labeling and clone formation were performed to evaluate the proliferative capacity of cells in these groups. The invasive and migratory abilities of cells in the KIF26B and si-KIF26B groups were assessed using Transwell assay. Additionally, the influence of KIF26B on the glycogen synthase kinase (GSK)-3ß/ß-catenin pathway was investigated through Western blot analysis. RESULTS: According to the results of RT-qPCR and Western blot analyses, the expression of KIF26B was predominantly higher in OSCC tissues compared to normal tissues (p < 0.01). Overexpression of KIF26B notably accelerated cell migration, invasion, and proliferation (p < 0.01), whereas knockdown of KIF26B significantly inhibited these processes (p < 0.01). Additionally, KIF26B overexpression led to increased levels of active ß-catenin, p-GSK-3, and c-myc (p < 0.01), while KIF26B silencing decreased the levels of these proteins (p < 0.01). CONCLUSION: Our findings suggest that KIF26B may play a role in the pathogenesis and progression of OSCC as an oncogene. This study establishes a foundation for the identification of potential therapeutic targets for OSCC.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Células Escamosas , Proliferación Celular , Cinesinas , Neoplasias de la Boca , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Línea Celular Tumoral , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Proliferación Celular/genética , Femenino , Masculino , Persona de Mediana Edad , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Movimiento Celular/genética , Anciano , Vía de Señalización Wnt/genética , beta Catenina/metabolismo , beta Catenina/genética
2.
Clin Chim Acta ; 560: 119717, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38782157

RESUMEN

BACKGROUND: Preeclampsia (PE) is a leading cause of maternal and fetal morbidity and mortality, with limited effective clinical treatment options. Active metabolomics offers a promising approach to uncover metabolic changes in PE and identify potential biomarkers or therapeutic targets. This study performed untargeted metabolomics using LC-MS to compare serum samples from preeclampsia and normal pregnancies. METHODS: We performed untargeted metabolomics using liquid chromatography-mass spectrometry (LC-MS) to compare serum samples from PE patients and normal pregnancies. We analyzed the alterations in metabolites and conducted functional experiments to assess the effects of LysoPE(16:0) on trophoblast cell invasion and migration. Mechanistic studies were performed to explore the potential targeting of GSK-3ß by LysoPE(16:0). RESULTS: Our metabolomics analysis revealed significant alterations in several metabolites, including lysophosphatidylcholines and organic acids. Notably, LysoPE(16:0) was found to be downregulated in the serum of PE patients. Functional experiments demonstrated that LysoPE(16:0) could promote trophoblast cell invasion and migration. Mechanistic studies suggest that the protective effect of LysoPE(16:0) against PE might be mediated through the modulation of the GSK-3ß/ß-Catenin pathway, with LysoPE(16:0) potentially targeting the GSK-3ß protein. CONCLUSIONS: Our findings highlight the potential role of LysoPE(16:0) in the pathophysiology of PE and its ability to modulate the GSK-3ß/ß-Catenin pathway. These results provide new insights into the metabolic changes associated with PE and suggest that LysoPE(16:0) could serve as a promising biomarker or therapeutic target for the prevention and treatment of PE.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta , Metabolómica , Preeclampsia , Humanos , Preeclampsia/sangre , Preeclampsia/metabolismo , Preeclampsia/diagnóstico , Preeclampsia/prevención & control , Femenino , Embarazo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Adulto , Trofoblastos/metabolismo , Movimiento Celular , Cromatografía Liquida
3.
Int J Nanomedicine ; 19: 2957-2972, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38549840

RESUMEN

Introduction: Nano-mesoporous bioactive glass and RGD peptide-coated collagen membranes have great potential in wound healing. However, the application of their compound has not been further studied. Our purpose is to prepare a novel bioactive collagen scaffold containing both NMBG stent and adhesion peptides (BM), which then proves its promising prospect the assessment of physical properties, biocompatibility, GSK-3ß/ß-catenin signaling axis and toxicological effects. Methods: The structural and morphological changes of BM were analyzed using scanning electron microscopy (SEM) and Energy Dispersive Spectroscopy (EDS). In vivo, wound healing of BM was assessed in SD rats through dynamic monitoring and calculation of wound healing rate. Immunohistofluorescence (IHF), H&E, and Masson staining were utilized; in vitro, primary cell culture, and a variety of assays including CCK-8, Transwell, Scratch, Immunocytofluorescence (ICF), and Western blot (WB) were performed, both for morphology and molecular analysis. Results and Discussion: Preparation of BM involved attaching NMBG to RGD-exposed collagen while avoiding the use of toxic chemical reagents. BM exhibited a distinctive superficial morphology with increased Si content, indicating successful NMBG attachment. In vivo studies on SD rats demonstrated the superior wound healing capability of BM, as evidenced by accelerated wound closure, thicker epithelial layers, and enhanced collagen deposition compared to the NC group. Additionally, BM promoted skin fibroblast migration and proliferation, possibly through activation of the GSK-3ß/ß-catenin signaling axis, which was crucial for tissue regeneration. This study underscored the potential of BM as an effective wound-healing dressing. Conclusion: A new method for synthesizing ECM-like membranes has been developed using nano-mesoporous bioactive glass and collagen-derived peptides. This approach enhances the bioactivity of biomaterials through surface functionalization and growth factor-free therapy.


Asunto(s)
Biomimética , beta Catenina , Ratas , Animales , Glucógeno Sintasa Quinasa 3 beta/metabolismo , beta Catenina/metabolismo , Ratas Sprague-Dawley , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos , Proliferación Celular , Péptidos/farmacología
4.
Ann Med Surg (Lond) ; 86(3): 1376-1385, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38463106

RESUMEN

Background: Some studies have found that the application of traditional Chinese medicine in the treatment of lung cancer has achieved satisfying results. Polyphyllin Ⅲ (PP Ⅲ) is a natural steroid saponin from P. polyphylla var. yunnanensis, and its analogs have played a wide role in anticancer research. This study aimed to investigate the effect of PP Ⅲ on the development of lung cancer and its molecular mechanism. Methods: A549 and NCI-H1299 cell lines were treated with PP Ⅲ in gradient concentration to detect the IC50 of the cells, and the optimal concentration was selected for subsequent experiments. The effects of PP III treatment on lung cancer were investigated in vitro and in vivo. Results: In vitro experiments, it was found that the proliferation, invasion, migration, and colony formation ability of cancer cells were significantly reduced after PP III treatment, while accompanied by a large number of cell apoptosis. Further detection showed that N-cadherin was significantly decreased, E-cadherin was increased, and Snail and Twist were decreased in A549 cells and NCI-H1299 cells, respectively. In addition, GSK-3ß expression was increased, while ß-catenin expression was reduced with PP III treatment. In the mouse model, it was demonstrated that the volume of transplanted tumors was significantly reduced after PP Ⅲ treatment. Conclusions: PP Ⅲ has the capacity to inhibit the progression of lung cancer and regulate epithelial-mesenchymal transition through the GSK-3ß/ß-catenin pathway to suppress the malignant behavior of cancer cells. The application of PP Ⅲ is expected to be an effective method for the treatment of lung cancer.

5.
Exp Ther Med ; 26(5): 514, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37840566

RESUMEN

Oral squamous cell carcinoma (OSCC) is one of the most common malignancies worldwide, which is associated with a poor prognosis. The present study aimed to investigate the role of cancerous inhibitor of protein phosphatase 2A (CIP2A) in OSCC and its regulatory effect on AKT1. Firstly, CIP2A and AKT1 expression in OSCC cells was detected by western blotting. After silencing CIP2A, cell viability and cell proliferation were assessed using the Cell Counting Kit-8 assay and 5-ethynyl-2'-deoxyuridine staining. Cell apoptosis was evaluated by TUNEL staining and the expression of apoptosis-related proteins was assessed using western blotting. Wound healing, Transwell and tube formation assays were performed to evaluate CAL-27 cell migration, invasion and human umbilical vein endothelial cell (HUVEC) tube formation. The interaction between CIP2A and AKT1 was identified by co-immunoprecipitation (co-IP). In addition, AKT1 was overexpressed in CIP2A-silenced CAL-27 cells to perform rescue experiments to analyze the malignant biological functions of CAL-27 cells. Finally, the expression of proteins in the glycogen synthase kinase (GSK)-3ß/ß-catenin pathway was determined by western blot analysis. Markedly elevated CIP2A and AKT1 expression was observed in OSCC cells. CIP2A knockdown inhibited the viability, proliferation, migration and invasion, and promoted the apoptosis of CAL-27 cells. Concurrently, CIP2A loss-of-function attenuated tube formation. Results of Co-IP confirmed there was an interaction between CIP2A and AKT1. Rescue experiments suggested that AKT1 overexpression alleviated the inhibitory effects of CIP2A knockdown on the viability, proliferation, migration and invasion of CAL-27 cells, as well as tube formation in HUVECs . Additionally, CIP2A silencing significantly downregulated phosphorylated-GSK-3ß and ß-catenin expression, which was reversed by AKT1 overexpression. In conclusion, CIP2A could interact with AKT1 to promote the malignant biological behaviors of OSCC cells by upregulating the GSK-3ß/ß-catenin pathway. These findings may provide a targeted therapy for OSCC treatment.

6.
Clin Exp Immunol ; 214(1): 120-129, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-37402316

RESUMEN

Excessive inflammatory injury is the main cause of the incidence of severe neonatal pneumonia (NP) and associated deaths. Although dickkopf-3 (DKK3) exhibits anti-inflammatory activity in numerous pathological processes, its role in NP is still unknown. In this study, human embryonic lung WI-38 and MRC-5 cells were treated with lipopolysaccharide (LPS) to induce inflammatory injury of NP in vitro. The expression of DKK3 was downregulated in LPS-stimulated WI-38 and MRC-5 cells. DKK3 overexpression decreased LPS-induced inhibition of cell viability, and reduced LPS-induced apoptosis of WI-38 and MRC-5 cells. DKK3 overexpression also reduced LPS-induced production of pro-inflammatory factors such as ROS, IL-6, MCP-1, and TNF-α. Nuclear respiratory factors 1 (NRF1) knockdown was found to upregulate DKK3 and inactivate the GSK-3ß/ß-catenin pathway in LPS-injured WI-38 and MRC-5 cells. NRF1 knockdown also suppressed LPS-induced inhibition on cell viability, repressed LPS-induced apoptosis, and inhibited the accumulation of ROS, IL-6, MCP-1, and TNF-α in LPS-injured WI-38 and MRC-5 cells. DKK3 knockdown or re-activation of the GSK-3ß/ß-catenin pathway reversed the inhibitory effects of NRF1 knockdown on LPS-induced inflammatory injury. In conclusion, NRF1 knockdown can alleviate LPS-triggered inflammatory injury by regulating DKK3 and the GSK-3ß/ß-catenin pathway.


Asunto(s)
Neumonía , Transducción de Señal , Recién Nacido , Humanos , Lipopolisacáridos , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/farmacología , beta Catenina/genética , beta Catenina/metabolismo , beta Catenina/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/genética , Proteínas Adaptadoras Transductoras de Señales/genética
7.
Biomol Ther (Seoul) ; 31(5): 515-525, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37366053

RESUMEN

The most common heart valve disorder is calcific aortic valve stenosis (CAVS), which is characterized by a narrowing of the aortic valve. Treatment with the drug molecule, in addition to surgical and transcatheter valve replacement, is the primary focus of researchers in this field. The purpose of this study is to determine whether niclosamide can reduce calcification in aortic valve interstitial cells (VICs). To induce calcification, cells were treated with a pro-calcifying medium (PCM). Different concentrations of niclosamide were added to the PCM-treated cells, and the level of calcification, mRNA, and protein expression of calcification markers was measured. Niclosamide inhibited aortic valve calcification as observed from reduced alizarin red s staining in niclosamide treated VICs and also decreased the mRNA and protein expressions of calcification-specific markers: runt-related transcription factor 2 and osteopontin. Niclosamide also reduced the formation of reactive oxygen species, NADPH oxidase activity and the expression of Nox2 and p22phox. Furthermore, in calcified VICs, niclosamide inhibited the expression of ß-catenin and phosphorylated glycogen synthase kinase (GSK-3ß), as well as the phosphorylation of AKT and ERK. Taken together, our findings suggest that niclosamide may alleviate PCM-induced calcification, at least in part, by targeting oxidative stress mediated GSK-3ß/ß-catenin signaling pathway via inhibiting activation of AKT and ERK, and may be a potential treatment for CAVS.

8.
Chem Biol Interact ; 368: 110218, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36223831

RESUMEN

Silica is a very common and important chemical raw material with a wide range of uses. Long-term inhalation of silica particles could cause lung toxicity, of which the most important representative is silicosis. Silicosis is a serious and fatal occupational pulmonary disease, characterized by persistent pulmonary inflammation and fibrosis. Despite intensive research, the toxic mechanism of silicosis caused by silica particles is not completely clear, which deserves further research and exploration. Many studies have indicated that the epithelial cells partially participate in the formation, accumulation, and activation of fibroblasts through epithelial-mesenchymal transition (EMT), which is conducive to the occurrence of fibrosis. Galectin-3 (Gal-3), widely expressed in epithelial cells, was observed to induce EMT in fibrotic diseases and tumors by regulating the GSK-3ß and ß-catenin. Previous studies have demonstrated that silica particles is indeed involved in the silicosis process by inducing EMT. However, it remains to be further studied whether there is a certain relationship between silica particles and Gal-3 expression, and whether Gal-3 also mediates the development of the silica particles-induced silicosis by regulating GSK-3ß/ß-catenin signal pathway-mediated EMT. Our research results showed that silica particles could significantly induce Gal-3 expression to promote the development of EMT through activating the GSK-3ß/ß-catenin signal pathway in mice and in A549 cells, which then significantly exacerbated the pulmonary fibrosis caused by silica particles. And the inhibition of Gal-3 could effectively inhibit the occurrence of EMT, and then effectively alleviate silicosis caused by silica particles. These findings would help us to further clarify the toxicological mechanisms of silicosis caused by silica particles and provide a novel target for prevention and intervention of silicosis.


Asunto(s)
Transición Epitelial-Mesenquimal , Silicosis , Ratones , Animales , beta Catenina/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Dióxido de Silicio/toxicidad , Galectina 3/metabolismo , Silicosis/prevención & control , Transducción de Señal , Fibrosis
9.
Heliyon ; 8(8): e10379, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36061014

RESUMEN

Background: Orthodontic tooth movement mainly depends on biological and mechanical reactions in the periodontium, such as the indispensable reconstruction process of the periodontal ligament and alveolar bone. To explore whether orthodontic compressive force can induce bone resorption during orthodontic tooth movement by regulating the GSK-3ß/ß-catenin pathway. Methods: We established orthodontic tooth movement models in Sprague-Dawley rats. In addition, compressive force-induced bone resorption that occurred during orthodontic tooth movement was analyzed by HE staining and micro-CT. The number and distribution of osteoclasts were observed by TRAP staining. Furthermore, pressure-induced bone resorption mediated by the GSK-3ß/ß-catenin signaling pathway was analyzed by immunohistochemistry. Results: As shown by the micro-CT results, bone parameters, such as bone mineral density (BMD), the bone volume fraction (BV/TV), and trabecular thickness (Tb. Th), were significantly decreased under orthodontic compressive force stimulation, in contrast with the dramatically increased trabecular spacing (Tb. Sp). During the process of tooth movement, the compressive force can induce bone resorption on the side with the force, which increases the expression of phosphorylated Ser-GSK-3ß and activation of the ß-catenin signaling pathway. Additionally, downregulation of the GSK-3ß activity further caused the downregulation of bone parameters, leading to bone loss. The TRAP staining and immunohistochemistry staining results indicated that orthodontic compressive force influenced osteoclast formation and the secretion of osteoclast-related cytokines, matrix metallopeptidase 9 (MMP-9) and receptor activator of nuclear factor-κB ligands (RANKLs), which is also related to the duration of orthodontic force. Conclusions: These results indicated that the GSK-3ß inhibitor can promote osteoclast formation on the side with orthodontic compressive force. In addition, the activation of the GSK-3ß/ß-catenin signaling pathway contributes to bone reconstruction caused by orthodontic compressive force. Therefore, the GSK-3ß/ß-catenin signaling pathway can be a potential target for further clinical applications.

10.
Front Cardiovasc Med ; 9: 970745, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36082129

RESUMEN

The mammalian heart possesses entire regeneration capacity after birth, which is lost in adulthood. The role of the kinase network in myocardial regeneration remains largely elusive. SGK3 (threonine-protein kinase 3) is a functional kinase we identified previously with the capacity to promote cardiomyocyte proliferation and cardiac repair after myocardial infarction. However, the upstream signals regulating SGK3 are still unknown. Based on the quantitative phosphoproteomics data and pulldown assay, we identified cyclin-dependent kinase 9 (CDK9) as a novel therapeutic target in regeneration therapy. The direct combination between CDK9 and SGK3 was further confirmed by co-immunoprecipitation (Co-IP). CDK9 is highly expressed in the newborn period and rarely detected in the adult myocardium. In vitro, the proliferation ratio of primary cardiomyocytes was significantly elevated by CDK9 overexpression while inhibited by CDK9 knockdown. In vivo, inhibition of CDK9 shortened the time window of cardiac regeneration after apical resection (AR) in neonatal mice, while overexpression of CDK9 significantly promoted mature cardiomyocytes (CMs) to re-enter the cell cycle and cardiac repair after myocardial infarction (MI) in adult mice. Mechanistically, CDK9 promoted cardiac repair by directly activating SGK3 and downstream GSK-3ß/ß-catenin pathway. Consequently, our study indicated that CDK9 might be a novel target for MI therapy by stimulating myocardial regeneration.

11.
Stem Cells Dev ; 30(16): 806-815, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34088227

RESUMEN

Mesenchymal stem cells (MSCs) have been shown to be involved in bone injury repair. Programmed cell death 4 (PDCD4) is not only a tumor suppressor gene but also plays roles in the regulation of MSC function. The aim of the study was to uncover PDCD4 potential regulatory roles and mechanisms in the osteogenic differentiation and bone defect repair of MSCs. shRNA technique was used to knock down PDCD4 expression in umbilical cord-derived mesenchymal stem cells (shPDCD4-UCMSCs). Their phenotype was characterized by flow cytometry and the differentiation potential was verified. We found that PDCD4 knockdown did not affect the surface molecule expression of UCMSCs, but significantly enhanced their osteogenic differentiation and osteogenesis-related molecule expression. Mechanistically, glycogen synthase kinase-3ß (GSK-3ß) phosphorylation and ß-catenin expression were significantly increased in shPDCD4-UCMSCs during the osteogenic differentiation process. The ß-catenin inhibitor PNU-74654 reversed shPDCD4-increased osteogenesis and osteogenesis-related molecule expression. The results of animal experiments showed that shPDCD4-UCMSCs markedly improved the defect healing in rabbits. Our findings suggest that PDCD4 acts as a negative regulator of MSC osteogenic differentiation through GSK-3ß/ß-catenin pathway. Targeting PDCD4 may be a way to improve MSC-mediated therapeutic effects on bone injury.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Glucógeno Sintasa Quinasa 3 beta , Células Madre Mesenquimatosas , Osteogénesis , Proteínas de Unión al ARN , beta Catenina , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Diferenciación Celular , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/genética , Proteínas de Unión al ARN/metabolismo , Conejos , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
12.
Stem Cell Reports ; 16(6): 1568-1583, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34019818

RESUMEN

Photobiomodulation therapy (PBMT) has shown encouraging results in the treatment of hair loss. However, the mechanism by which PBMT controls cell behavior to coordinate hair cycle is unclear. Here, PBMT is found to drive quiescent hair follicle stem cell (HFSC) activation and alleviate hair follicle atrophy. Mechanistically, PBMT triggers a new hair cycle by upregulating ß-CATENIN expression in HFSCs. Loss of ß-Catenin (Ctnnb1) in HFSCs blocked PBMT-induced hair regeneration. Additionally, we show PBMT-induced reactive oxygen species (ROS) activate the PI3K/AKT/GSK-3ß signaling pathway to inhibit proteasome degradation of ß-CATENIN in HFSCs. Furthermore, PBMT promotes the expression and secretion of WNTs in skin-derived precursors (SKPs) to further activate the ß-CATENIN signal in HFSCs. By contrast, eliminating ROS or inhibiting WNT secretion attenuates the activation of HFSCs triggered by PBMT. Collectively, our work suggests that PBMT promotes hair regeneration through synergetic activation of ß-CATENIN in HFSCs by ROS and paracrine WNTs by SKPs.


Asunto(s)
Alopecia/terapia , Folículo Piloso/metabolismo , Terapia por Luz de Baja Intensidad/métodos , Especies Reactivas de Oxígeno/metabolismo , Células Madre/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Femenino , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes/métodos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Regeneración
13.
Life Sci ; 262: 118491, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33011219

RESUMEN

AIMS: Aurora kinase A (AURKA) is a mitotic serine/threonine kinase that contributes to the regulation of cell-cycle progression. AURKA has been shown to further enhance Wnt/ß-catenin signaling; however, in the context of driving aortic-dissecting aneurysm (ADA), the molecular details of this phenomenon remain poorly understood. MATERIALS AND METHODS: A total of 43 specimens of ADA tissues and eleven healthy aortic tissues as controls were collected from the hospital. Pathological changes were observed by hematoxylin and eosin staining. AURKA expression in aortic tissues was detected by real-time quantitative polymerase chain reaction (RT-qPCR), western blot, and immunohistochemistry staining. The proliferative and migratory effects of AURKA were observed in cultured vascular smooth muscle cells (VSMCs). KEY FINDINGS: AURKA expression was significantly increased in aorta samples from ADA patients relative to those from normal donors, and the expression was even higher in ruptured ADA tissues. AURKA overexpression promoted and AURKA knockdown inhibited, respectively, the proliferation, and migration of VSMCs. Angiotensin II (AngII) treatment of VSMCs significantly increased AURKA expression. The knockdown of AURKA partially reversed AngII-induced VSMC proliferation and migration. Finally, the downregulation of AURKA inhibited cell proliferation by inactivating the p-GSK-3ß/ß-catenin pathway. SIGNIFICANCE: The GSK-3ß/ß-catenin signaling pathway participates in the AURKA-regulated proliferation and migration of VSMCs. The expression of AURKA may be involved in the phenotypic conversion of VSMC and the occurrence and development of ADA and could be a potential molecular target for ADA therapy.


Asunto(s)
Disección Aórtica/fisiopatología , Aurora Quinasa A/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Músculo Liso Vascular/citología , Adulto , Anciano , Disección Aórtica/genética , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Regulación hacia Abajo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Persona de Mediana Edad , Miocitos del Músculo Liso/metabolismo , Regulación hacia Arriba , Vía de Señalización Wnt/genética , Adulto Joven
14.
Environ Toxicol Pharmacol ; 79: 103432, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32502517

RESUMEN

BACKGROUND: Diabetic peripheral neuropathy, a common complication of diabetic mellitus, has brought a threaten on patients' health. The bone marrow-derived mesenchymal stem cells (BMSCs) were reported to play an important role in diverse diseases. Nevertheless, the specific function of BMSCs in diabetic peripheral neuropathy remained uncharacterized. METHODS: A wide range of experiments including RT-qPCR, western blot, H&E staining, oxidative stress assessment, measurement of thermal sensitivity, ELISA, urine protein and CCK-8 assays were implemented to explore the function and mechanism of BMSCs in vivo and vitro. RESULTS: The experimental results displayed that BMSCs improve STZ-induced diabetes symptoms in rats by decreasing blood glucose and urinary protein. Functionally, BMSCs ameliorate oxidative stress, painful diabetic neuropathy, neurotrophic status and angiogenesis in STZ-induced rats. Moreover, BMSCs participate in the regulation of sciatic neuro morphology in diabetic neuropathy rat model. In mechanism, BMSCs alleviate diabetic peripheral neuropathy via activating GSK-3ß/ß-catenin signaling pathway in rats and improve Schwann's cells viability by activating GSK-3ß/ß-catenin signaling pathway under high glucose. CONCLUSIONS: We verified that BMSCs alleviate diabetic peripheral neuropathy of rats induced by STZ via activating GSK-3ß/ß-catenin signaling pathway, which implied a novel biomarker for diabetic peripheral neuropathy treatment.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Neuropatías Diabéticas/terapia , Trasplante de Células Madre Mesenquimatosas , Animales , Glucemia , Peso Corporal , Células de la Médula Ósea/citología , Supervivencia Celular , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Neuropatías Diabéticas/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Masculino , Estrés Oxidativo , Ratas Sprague-Dawley , Células de Schwann , beta Catenina/metabolismo
15.
Cell Signal ; 71: 109570, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32084532

RESUMEN

BACKGROUND: Pathological retinal neovascularization is a disease characterized by abnormal angiogenesis in retina that is a major cause of blindness in humans. Previous reports have highlighted the involvement of microRNAs (miRNAs) in retinal angiogenesis. Therefore, we aimed at exploring the mechanism underlying miR-203 regulating the progression of pathological retinal neovascularization. METHODS: Initially, the mouse model of pathological retinal neovascularization disease was established and the hypoxia-induced human retinal microvascular endothelial cells (HRMECs) were generated. Then, miR-203 and SNAI2 expression in HRMECs and retinal tissues was examined. Subsequently, the effects of miR-203 and SNAI2 on viability, migration, apoptosis and angiogenesis of HRMECs were investigated, with the expression of Bax, Ki-67, MMP-2, MMP-9, VEGF and CD34 measured. Finally, the regulation of miR-203 or SNAI2 on GSK-3ß/ß-catenin pathway was determined through examining the levels of phosphorylated p-GSK-3ß and ß-catenin. RESULTS: Poorly expressed miR-203 and highly expressed SNAI2 were found in HRMECs and retinal tissues of pathological retinal neovascularization. Importantly, overexpressed miR-203 or silencing SNAI2 inhibited viability, migration and angiogenesis but promoted apoptosis of HRMECs, evidenced by elevated Bax expression but reduced expression of Ki-67, MMP-2, MMP-9, VEGF and CD34. Moreover, overexpression of miR-203 was found to repress the GSK-3ß/ß-catenin pathway by downregulating SNAI2. CONCLUSION: Collectively, this study demonstrated that overexpression of miR-203 suppressed the angiogenesis in mice with pathological retinal neovascularization disease via the inactivation of GSK-3ß/ß-catenin pathway by inhibiting SNAI2, which provided a novel therapeutic insight for pathological retinal neovascularization disease.


Asunto(s)
Regulación hacia Abajo/genética , MicroARNs/metabolismo , Neovascularización Retiniana/genética , Factores de Transcripción de la Familia Snail/genética , Animales , Apoptosis/genética , Secuencia de Bases , Hipoxia de la Célula/genética , Movimiento Celular/genética , Supervivencia Celular/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Ratones Endogámicos C57BL , MicroARNs/genética , Microvasos/metabolismo , Retina/patología , Transducción de Señal , Factores de Transcripción de la Familia Snail/metabolismo , Regulación hacia Arriba/genética , beta Catenina/metabolismo
16.
Phytomedicine ; 59: 152759, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31004883

RESUMEN

BACKGROUND: Pseudolaric acid B (PAB), a diterpene acid isolated from the root bark of Pseudolarix kaempferi, exhibits a potent anti-cancer activity in a variety of tumor cells. PURPOSE: The present study was designed to evaluate the anti-cancer effects of PAB on hepatocellular carcinoma (HCC) cell lines in vitro, and to explore the underlying mechanism. METHODS: The anti-proliferative activity of PAB on HCC cells were assessed via sulforhodamine B staining, colony formation, cell cycle analysis, respectively. Apoptosis was detected using Annexin V/propidium iodide double staining and diamidino-phenyl-indole staining, respectively. Protein expression regulated by PAB treatment was tested by western blotting. RESULTS: The present results showed that PAB significantly inhibited the proliferation of HepG2, SK-Hep-1, and Huh-7 HCC cell lines in vitro with IC50 values of 1.58, 1.90, and 2.06 µM, respectively. Furthermore, PAB treatment repressed the colony formation in HepG2, SK-Hep-1, and Huh-7 HCC cell lines. Flow cytometry analysis revealed that PAB caused an obvious cell cycle arrest in G2/M phase and induced apoptosis with the induction of p21, Bax, cleaved-caspase-3, and cleaved-PARP in human HepG2 and SK-Hep-1 cells. Mechanistically, PAB treatment down-regulated the phosphorylation of STAT3, ERK1/2, and Akt. Moreover, abnormal GSK-3ß/ß-catenin signaling in HepG2 cells was remarkably suppressed by PAB treatment. Finally, proliferation markers including cyclin D1 and c-Myc, and anti-apoptosis proteins such as Bcl-2 and survivin were also down-regulated by PAB treatment in HepG2 cells. CONCLUSION: Taken together, our results suggest that PAB exerts anti-cancer activity in HCC cells through inhibition of STAT3, ERK1/2, Akt, and GSK-3ß/ß-catenin carcinogenic signaling pathways, and may be used as a phytomedicine in the treatment of HCC.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Carcinogénesis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Diterpenos/farmacología , Neoplasias Hepáticas/metabolismo , Hígado/efectos de los fármacos , Extractos Vegetales/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Diterpenos/uso terapéutico , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Hep G2 , Humanos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Pinaceae/química , Transducción de Señal/efectos de los fármacos
17.
J Exp Clin Cancer Res ; 37(1): 122, 2018 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-29940988

RESUMEN

BACKGROUND: Serum and glucocorticoid-regulated kinase 3 (SGK3) has been reported to play an important role in tumour progression, but its role in cancer stem cells (CSCs) remains obscure. The phosphoinositide 3-kinase (PI3K) pathway is considered a hallmark of cancer. Although many PI3K pathway-targeted therapies have been tested in oncology trials, the results are not satisfactory. METHODS: We used spheroids cultured in serum-free culture medium and MicroBead isolation to obtain liver CSCs. Spheroid formation assay and flow cytometric analysis were performed to investigate liver CSC expansion. Real-time polymerase chain reaction (PCR), western blot and immunofluorescence were used to assess gene expression in cell lines. RESULTS: We found that SGK3 is preferentially activated in liver CSCs. Upregulated SGK3 significantly increases the expansion of liver CSCs. Conversely, suppression of SGK3 in human hepatocarcinoma (HCC) cells had an opposite effect. Mechanistically, SGK3 promoted ß-catenin accumulation by suppressing GSK-3ß-mediated ß-catenin degradation in liver CSCs, and then promoting the expansion of liver CSCs. Prolonged treatment of HCC cells with class I PI3K inhibitors leads to activation of SGK3 and expansion of liver CSCs. Inhibition of hVps34 can block SGK3 activity and suppress liver CSC expansion induced by PI3K inhibitors. More importantly, we also found that prolonged treatment of HCC cells with PI3K inhibitors stimulates the ß-catenin signalling pathway via activation of SGK3. CONCLUSIONS: Prolonged inhibition of class I PI3K promotes liver CSC expansion by augmenting SGK3-dependent ß-catenin stabilisation, and effective inhibition of SGK3 signalling may be useful in eliminating liver CSCs and in PI3K pathway-targeted cancer therapies.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Transducción de Señal/efectos de los fármacos , beta Catenina/metabolismo , Animales , Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Modelos Biológicos , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Int J Biol Sci ; 14(5): 497-507, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29805301

RESUMEN

Background: It has been reported that ultrasound enhances peripheral nerve regeneration, but the mechanism remains elusive. Low-intensity pulsed ultrasound (LIPUS) has been reported to enhance proliferation and alter protein production in various types of cells. In this study, we detected the effects of LIPUS on Schwann cells. Material and methods: Schwann cells were separated from new natal Sprague-Dawley rat sciatic nerves and were cultured and purified. The Schwann cells were treated by LIPUS for 10 minutes every day, with an intensity of 27.37 mW/cm2. After treatment for 5 days, MTT, EdU staining, and flow cytometry were performed to examine cell viability and proliferation. Neurotrophic factors, including FGF, NGF, BDNF, and GDNF, were measured by western blot and real-time PCR. GSK-3ß, p-GSK-3ß, ß-catenin and Cyclin D1 protein levels were detected using a western blot analysis. The expression of Cyclin D1 was also detected by immunofluorescence. Results: MTT and EdU staining showed that LIPUS increased the Schwann cells viability and proliferation. Compared to the control group, LIPUS increased the expression of growth factors and neurotrophic factors, including FGF, NGF, BDNF, GDNF, and Cyclin D1. Meanwhile, GSK-3ß activity was inhibited in the LIPUS group as demonstrated by the increased level of p-GSK-3ß and the ratio of the p-GSK-3ß/GSK-3ß level. The mRNA and protein expressions of ß-catenin were increased in the LIPUS group. However, SB216763, a GSK-3ß inhibitor, reversed the effects of LIPUS on Schwann cells. Conclusion: LIPUS promotes Schwann cell viability and proliferation by increasing Cyclin D1 expression via enhancing the GSK-3ß/ß-catenin signaling pathway.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células de Schwann/citología , Ondas Ultrasónicas , beta Catenina/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Ciclina D1/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Ciático/patología , Transducción de Señal
19.
IUBMB Life ; 70(3): 224-236, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29389061

RESUMEN

Dysregulation of microRNA expression plays a pivotal role in the initiation and progression of a variety of human carcinomas including prostate cancer. Our previous studies have demonstrated that the silence of miR-203 contributes to the invasiveness of malignant breast cancer cells by targeting SNAI2. However, the effects and underlying mechanisms of miR-203/SNAI2 axis in prostate cancer have not been elucidated. The aim of this study is to explore the effects of miR-203/SNAI2 axis on the biological characteristics of prostate carcinomas both in vitro and in vivo. We found that miR-203 was significantly downregulated in prostate cancer cell lines compared with immortalized prostate epithelial cells using semi-quantitative PCR and real-time PCR, as well as in clinical prostate cancer tissues compared to normal tissues using TCGA analysis. Functionally, miR-203 inhibited prostate cancer cell proliferation, migration, endothelial cell tube formation and cancer stemness in vitro. Meanwhile, overexpression of miR-203 suppressed SNAI2 expression both in DU145 and PC3 cells. In addition, the in vivo study showed that miR-203 suppressed tumorigenicity, metastasis and angiogenesis of DU145 cells. Ectopic expression of SNAI2 rescued the inhibitory effects of miR-203 both in vitro and in vivo. Importantly, the EMT markers CDH1 and VIMENTIN were modulated by the miR-203/SNAI2 axis. Furthermore, the GSK-3ß/ß-CATENIN signal pathway was suppressed by miR-203 and could be reactivated by SNAI2. Taken together, this research unveiled the function of miR-203/SNAI2 axis in tumorigenesis, angiogenesis, stemness, metastasis and GSK-3ß/ß-CATENIN signal pathway in prostate cancer and gave insights into miR-203/SNAI2-targeting therapy for prostate cancer patients. © 2018 IUBMB Life, 70(3):224-236, 2018.


Asunto(s)
MicroARNs/genética , Neoplasias de la Próstata/genética , Factores de Transcripción de la Familia Snail/genética , beta Catenina/genética , Anciano , Animales , Apoptosis/genética , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Neovascularización Patológica/genética , Neoplasias de la Próstata/patología , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Mol Histol ; 49(1): 75-84, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29224185

RESUMEN

Orthodontic force-induced osteogenic differentiation and bone formation at tension sites play a critical role in orthodontic tooth movement. However, the molecular mechanism underlying this phenomenon is poorly understood. In the current study, we investigated the involvement of the GSK-3ß/ß-catenin signaling pathway, which is critical for bone formation during tooth movement. We established a rat tooth movement model to test the hypothesis that orthodontic force may stimulate bone formation at the tension site of the moved tooth and promote the rate of tooth movement via regulation of the GSK-3ß/ß-catenin signaling pathway. Our results showed that continued mechanical loading increased the distance between the first and second molar in rats. In addition, the loading force increased bone formation at the tension site, and also increased phospho-Ser9-GSK-3ß expression and ß-catenin signaling pathway activity. Downregulation of GSK-3ß activity further increased bone parameters, including bone mineral density, bone volume to tissue volume and trabecular thickness, as well as ALP- and osterix-positive cells at tension sites during tooth movement. However, ICG-001, the ß-catenin selective inhibitor, reversed the positive effects of GSK-3ß inhibition. In addition, pharmaceutical inhibition of GSK-3ß or local treatment with ß-catenin inhibitor did not influence the rate of tooth movement. Based on these results, we concluded that GSK-3ß/ß-catenin signaling contributes to the bone remodeling induced by orthodontic forces, and can be used as a potential therapeutic target in clinical dentistry.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Osteogénesis , Estrés Mecánico , Técnicas de Movimiento Dental , beta Catenina/metabolismo , Animales , Fenómenos Biomecánicos , Remodelación Ósea , Ratas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA